Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Nicole H. Smith is active.

Publication


Featured researches published by Nicole H. Smith.


Clinical & Developmental Immunology | 2012

Initiation and Regulation of Complement during Hemolytic Transfusion Reactions

Sean R. Stowell; Anne M. Winkler; Cheryl L. Maier; C. Maridith Arthur; Nicole H. Smith; Kathryn R. Girard-Pierce; Richard D. Cummings; James C. Zimring; Jeanne E. Hendrickson

Hemolytic transfusion reactions represent one of the most common causes of transfusion-related mortality. Although many factors influence hemolytic transfusion reactions, complement activation represents one of the most common features associated with fatality. In this paper we will focus on the role of complement in initiating and regulating hemolytic transfusion reactions and will discuss potential strategies aimed at mitigating or favorably modulating complement during incompatible red blood cell transfusions.


Transfusion | 2014

Strain-specific red blood cell storage, metabolism, and eicosanoid generation in a mouse model

James C. Zimring; Nicole H. Smith; Sean R. Stowell; Jill M. Johnsen; Lauren N. Bell; Richard O. Francis; Eldad A. Hod; Jeanne E. Hendrickson; John D. Roback; Steven L. Spitalnik

Red blood cell (RBC) transfusion is a lifesaving therapy, the logistic implementation of which requires RBC storage. However, stored RBCs exhibit substantial donor variability in multiple characteristics, including hemolysis in vitro and RBC recovery in vivo. The basis of donor variability is poorly understood.


Transfusion | 2013

Addition of ascorbic acid solution to stored murine red blood cells increases posttransfusion recovery and decreases microparticles and alloimmunization.

Sean R. Stowell; Nicole H. Smith; James C. Zimring; Xiaoyun Fu; Andre F. Palmer; Jorge Fontes; Uddyalok Banerjee; Mark H. Yazer

The storage of red blood cells (RBCs) results in numerous changes, which over time result in decreased recovery of transfused RBCs. In addition (at least in animal models), stored RBCs can be more immunogenic and also stimulate the systemic release of inflammatory cytokines in transfusion recipients. One component of the RBC storage lesion is the accumulation of oxidative damage. We tested the hypothesis that adding a chemical antioxidant (ascorbic acid) to stored RBCs would improve the quality of the stored RBCs.


Transfusion | 2014

Transfusion of murine red blood cells expressing the human KEL glycoprotein induces clinically significant alloantibodies

Sean R. Stowell; Kathryn R. Girard-Pierce; Nicole H. Smith; Kate L. Henry; C. Maridith Arthur; James C. Zimring; Jeanne E. Hendrickson

Red blood cell (RBC) alloantibodies to nonself antigens may develop after transfusion or pregnancy, leading to morbidity and mortality in the form of hemolytic transfusion reactions or hemolytic disease of the newborn. A better understanding of the mechanisms of RBC alloantibody induction, or strategies to mitigate the consequences of such antibodies, may ultimately improve transfusion safety. However, such studies are inherently difficult in humans.


Transfusion | 2012

Generation of transgenic mice with antithetical KEL1 and KEL2 human blood group antigens on red blood cells

Nicole H. Smith; Kate L. Henry; Chantel M. Cadwell; Ashley Bennett; Jeanne E. Hendrickson; Tom Frame; James C. Zimring

BACKGROUND: KEL1, also known as “K,” is one of the most immunogenic red blood cell (RBC) antigens. KEL2, also known as “k,” differs from KEL1 by a single amino acid. Anti‐Kell system antibodies can lead to significant adverse clinical outcomes in humans, including hemolytic complications in alloimmunized transfusion recipients or in infants of alloimmunized mothers. To provide a platform for in‐depth immunologic studies of alloimmunization and subsequent sequelae, we generated transgenic mice expressing the human KEL1 or KEL2 antigens.


Journal of Immunology | 2013

Antigen Modulation Confers Protection to Red Blood Cells from Antibody through Fcγ Receptor Ligation

Sean R. Stowell; Justine S. Liepkalns; Jeanne E. Hendrickson; Kathryn R. Girard-Pierce; Nicole H. Smith; C. Maridith Arthur; James C. Zimring

Autoantibodies and alloantibodies can damage self-tissue or transplanted tissues through either fixation of complement or ligation of FcγRs. Several pathways have been described that imbue self-tissues with resistance to damage from complement fixation, as a protective measure against damage from these Abs. However, it has been unclear whether parallel pathways exist to provide protection from FcγR ligation by bound Abs. In this article, we describe a novel pathway by which cell surface Ag is specifically decreased as a result of Ab binding (Ag modulation) to the extent of conferring protection to recognized cells from Fcγ-dependent clearance. Moreover, the Ag modulation in this system requires FcγR ligation. Together, these findings provide unique evidence of self-protective pathways for FcγR-mediated Ab damage.


Blood | 2013

Alloantibodies to a paternally derived RBC KEL antigen lead to hemolytic disease of the fetus/newborn in a murine model

Sean R. Stowell; Kate L. Henry; Nicole H. Smith; Krystalyn E. Hudson; Greg R. Halverson; Jaekeun Park; Ashley Bennett; Kathryn R. Girard-Pierce; C. Maridith Arthur; Silvia T. Bunting; James C. Zimring; Jeanne E. Hendrickson

Exposure to nonself red blood cell (RBC) antigens, either from transfusion or pregnancy, may result in alloimmunization and incompatible RBC clearance. First described as a pregnancy complication 80 years ago, hemolytic disease of the fetus and newborn (HDFN) is caused by alloimmunization to paternally derived RBC antigens. Despite the morbidity/mortality of HDFN, women at risk for RBC alloimmunization have few therapeutic options. Given that alloantibodies to antigens in the KEL family are among the most clinically significant, we developed a murine model with RBC-specific expression of the human KEL antigen to evaluate the impact of maternal/fetal KEL incompatibility. After exposure to fetal KEL RBCs during successive pregnancies with KEL-positive males, 21 of 21 wild-type female mice developed anti-KEL alloantibodies; intrauterine fetal anemia and/or demise occurred in a subset of KEL-positive pups born to wild type, but not agammaglobulinemic mothers. Similar to previous observations in humans, pregnancy-associated alloantibodies were detrimental in a transfusion setting, and transfusion-associated alloantibodies were detrimental in a pregnancy setting. This is the first pregnancy-associated HDFN model described to date, which will serve as a platform to develop targeted therapies to prevent and/or mitigate the dangers of RBC alloantibodies to fetuses and newborns.


Molecular Immunology | 2011

Alloimmunization against RBC or PLT Antigens Is Independent of TRIM21 Expression in a Murine Model

Seema R. Patel; Jeanne E. Hendrickson; Nicole H. Smith; Chantel M. Cadwell; Keiko Ozato; Herbert C. Morse; Ryusuke Yoshimi; James C. Zimring

Generation of alloantibodies to transfused RBCs can be a serious medical problem for patients who require chronic RBC transfusion therapy. Patients with sickle cell disease have a substantially increased rate of alloimmunization compared to other chronically transfused populations. A recent study has forwarded the hypothesis that a polymorphism in an immunoregulatory gene in close proximity to beta-globin (TRIM21 rs660) plays a role in the increased rates of RBC alloimmunization in sickle cell patients. In particular, it was hypothesized that rs660C/T decreases expression of TRIM21, resulting in loss of a negative feedback pathway in immune responses and increased RBC alloimmunization. To test the effects of TRIM21 expression on alloimmunization, we analyzed antibody responses to alloantigens on RBCs and platelets transfused into wild-type and TRIM21 KO mice. No significant increases were seen in the frequency or magnitude of humoral immunization to alloantigens on transfused RBCs or platelets in adult or juvenile TRIM21 KO recipients compared to wild-type controls. Moreover, recipient inflammation with poly (I:C) enhanced RBC alloimmunization to similar degrees in both TRIM21 KO and wild-type control recipients. Together, these data rule out the hypothesis that decreased TRIM21 expression enhances transfusion induced humoral alloimmunization, in the context of a reductionist murine model.


Journal of Immunology | 2017

Antigen Density Dictates Immune Responsiveness following Red Blood Cell Transfusion.

Connie M. Arthur; Seema R. Patel; Nicole H. Smith; Ashley Bennett; Nourine A. Kamili; Amanda Mener; Christian Gerner-Smidt; Harold C. Sullivan; J. Scott Hale; Andreas Wieland; Benjamin Alan Youngblood; James C. Zimring; Jeanne E. Hendrickson; Sean R. Stowell

Although RBC transfusion can result in the development of anti-RBC alloantibodies that increase the probability of life-threatening hemolytic transfusion reactions, not all patients generate anti-RBC alloantibodies. However, the factors that regulate immune responsiveness to RBC transfusion remain incompletely understood. One variable that may influence alloantibody formation is RBC alloantigen density. RBC alloantigens exist at different densities on the RBC surface and likewise exhibit distinct propensities to induce RBC alloantibody formation. However, although distinct alloantigens reside on the RBC surface at different levels, most alloantigens also represent completely different structures, making it difficult to separate the potential impact of differences in Ag density from other alloantigen features that may also influence RBC alloimmunization. To address this, we generated RBCs that stably express the same Ag at different levels. Although exposure to RBCs with higher Ag levels induces a robust Ab response, RBCs bearing low Ag levels fail to induce RBC alloantibodies. However, exposure to low Ag–density RBCs is not without consequence, because recipients subsequently develop Ag-specific tolerance. Low Ag–density RBC–induced tolerance protects higher Ag–density RBCs from immune-mediated clearance, is Ag specific, and occurs through the induction of B cell unresponsiveness. These results demonstrate that Ag density can potently impact immune outcomes following RBC transfusion and suggest that RBCs with altered Ag levels may provide a unique tool to induce Ag-specific tolerance.


American Journal of Transplantation | 2012

Mechanisms of alloimmunization and subsequent bone marrow transplantation rejection induced by platelet transfusion in a murine model.

Seema R. Patel; Nicole H. Smith; Linda M. Kapp; James C. Zimring

For many nonmalignant hematological disorders, HLA‐matched bone marrow transplantation (BMT) is curative. However, due to lack of neoplasia, the toxicity of stringent conditioning regimens is difficult to justify, and reduced intensity conditioning is used. Unfortunately, current reduced intensity regimens have high rates of BMT rejection. We have recently reported in a murine model that mHAs on transfused platelet products induce subsequent BMT rejection. Most nonmalignant hematological disorders require transfusion support prior to BMT and the rate of BMT rejection in humans correlates with the number of transfusions given. Herein, we perform a mechanistic analysis of platelet transfusion‐induced BMT rejection and report that unlike exposure to alloantigens during transplantation, platelet transfusion primes alloimmunity but does not stimulate full effector function. Subsequent BMT is itself an additional and distinct immunizing event, which does not induce rejection without antecedent priming from transfusion. Both CD4+ and CD8+ T cells are required for priming during platelet transfusion, but only CD8+ T cells are required for BMT rejection. In neither case are antibodies required for rejection to occur.

Collaboration


Dive into the Nicole H. Smith's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eldad A. Hod

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John D. Roback

Emory University Hospital

View shared research outputs
Top Co-Authors

Avatar

Richard O. Francis

Columbia University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge